Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
2.
Thromb Haemost ; 97(6): 965-73, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17549299

RESUMO

Parnaparin, a low-molecular-weight heparin (LMWH), prevents platelet activation and interaction with polymorphonuclear leukocyte (PMN) in a washed cell system. The in-vitro effect of parnaparin was studied here on platelet-PMN aggregates formed with more physiologic approaches in whole blood, in parallel with unfractionated heparin and enoxaparin, another LMWH. Citrated blood from healthy subjects was stimulated: i) from passage through the "Platelet Function Analyzer" (PFA-100), a device that exposes blood to standardized high shear flow through collagen/ADP cartridges; ii) by collagen and ADP (2 and 50 mug/ml, respectively) added in combination under stirring in an aggregometer cuvette; iii) with recombinant Tissue Factor, to generate thrombin concentrations able to activate platelets without inducing blood clotting, or iv) the Thrombin Receptor Activating Peptide-6 (TRAP-6). Platelet P-selectin and platelet-PMN aggregates were measured by flow cytometry upon stimulation of blood. Fibrinogen binding to platelets and markers of PMN activation were also detected. Platelet P-selectin expression and platelet-PMN aggregate formation were induced in all four activation conditions tested. Parnaparin prevented in a concentration-dependent manner (0.3-0.8 IUaXa/ml) the expression of P-selectin and the formation of mixed aggregates, while the two reference heparin preparations had a much weaker effect. Platelet fibrinogen binding and PMN activation markers (fibrinogen binding, CD11b and CD40) were also prevented by parnaparin. These data extend in more physiological systems of platelet activation, the anti-inflammatory profile of parnaparin, previously reported in washed cells. The greater effect of parnaparin, as compared to the reference heparins, could be due to chemico-physical differences possibly unrelated to their anticoagulant effect.


Assuntos
Anti-Inflamatórios/farmacologia , Plaquetas/efeitos dos fármacos , Heparina de Baixo Peso Molecular/farmacologia , Leucócitos/efeitos dos fármacos , Selectina-P/metabolismo , Adesividade Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Difosfato de Adenosina/metabolismo , Plaquetas/metabolismo , Antígeno CD11b/metabolismo , Antígenos CD40/metabolismo , Adesão Celular/efeitos dos fármacos , Colágeno/metabolismo , Relação Dose-Resposta a Droga , Enoxaparina/farmacologia , Fibrinogênio/metabolismo , Humanos , Técnicas In Vitro , Leucócitos/metabolismo , Fragmentos de Peptídeos/farmacologia , Testes de Função Plaquetária/instrumentação , Estresse Mecânico , Trombina/metabolismo , Tromboplastina/metabolismo
3.
Haematologica ; 90(6): 833-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15951297

RESUMO

BACKGROUND AND OBJECTIVES: Heparin might possess anti-thrombotic properties other than anticoagulation. The aim of the present study was to test the effects of a low-molecular weight heparin, parnaparin, on adhesive molecule-mediated platelet-polymorphonuclear (PMN) leukocyte interactions and on PMN function. DESIGN AND METHODS: Platelets and PMN were isolated from citrated blood from healthy subjects. Pre-activated platelets incubated with PMN under dynamic conditions formed mixed cell aggregates. In previous experiments PMN were stimulated in vitro by purified P-selectin or formyl-methionyl-leucyl-phenylalanine (fMLP). Dual color flow cytometry was used to detect the formation of platelet-PMN mixed cell aggregates, and PMN activation was tested for by measuring L-selectin shedding, tissue factor expression and PMN degranulation. The effect of parnaparin was compared to that of unfractionated heparin. RESULTS: Parnaparin, at a concentration of 0.3-0.8 IUaXa/mL, inhibited the formation of mixed cell aggregates (48.8+/-9.7% of total PMN population) by up to 60% in a concentration-dependent manner, while heparin inhibited aggregation up to 40%. Parnaparin, (0.3-0.8 IUaXa/mL), prevented L-selectin shedding from PMN, which was induced by purified P-selectin (5 mg/mL) or fMLP (0.5 mmol/L) by 65% and 67%, respectively. Inhibition was independent of incubation time (5-20 min). Parnaparin (0.8 IUaXa/mL) also inhibited tissue factor expression on PMN (% of positive cells), which was induced by P-selectin or fMLP (185+/-10 and 241+/-80% of basal value, respectively). Parnaparin protected PMN from degranulation after challenge with either stimulus (>95% inhibition). All the effects of parnaparin were observed with heparin at similar concentrations, although to a lesser extent and were often not significantly different from events in controls. INTERPRETATION AND CONCLUSIONS: In conclusion, the process of depolymerization of heparin to obtain low molecular weight parnaparin resulted in an increased, anticoagulant-independent effect on PMN function. Thus, the overall anti-thrombotic properties of parnaparin may be partly due to a leukocyte-mediated anti-inflammatory effect.


Assuntos
Plaquetas/citologia , Fibrinolíticos/farmacologia , Heparina de Baixo Peso Molecular/farmacologia , Neutrófilos/citologia , Anti-Inflamatórios/farmacologia , Adesão Celular , Citometria de Fluxo , Heparina/química , Humanos , Selectina L/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Selectina-P/metabolismo , Ativação Plaquetária , Fatores de Tempo
4.
FASEB J ; 17(9): 1171-3, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12709408

RESUMO

In addition to inhibiting formation of prothrombotic eicosanoids, aspirin causes the acetylation of cyclooxygenase (COX)-2. The acetylated COX-2 remains active, and upon cell activation, initiates the generation of 15R-HETE, a lipid substrate for 5-lipoxygenase (LOX) leading to the formation of 15-epi-LXA4 (also termed "aspirin-triggered lipoxin," or ATL). Because ATL potently inhibits polymorphonuclear cell (PMN) function, we assessed the relative contribution of this lipid mediator in conjunction with another 5-LOX product, the leukotriene (LT)B4, to the pathogenesis of acute damage and gastric adaptation to aspirin. Data presented herein indicate that acute injury and gastric adaptation to aspirin is associated with ATL generation. Administration of COX inhibitors (celecoxib, indomethacin, ketoprofen) to aspirin-treated rats exacerbated acute injury and abolished adaptation to aspirin. Moreover, it inhibited ATL formation and caused a four- to fivefold increase in LTB4 synthesis. In contrast, licofelone, a COX/5-LOX inhibitor, did not exacerbate acute gastric injury nor did it interfere with gastric adaptation to aspirin. Although licofelone blocked ATL and LTB4 formation in aspirin-treated rats, it attenuated aspirin-induced gastric PMN margination. These findings indicate that the balance between the production of LTB4 and ATL modulates PMN recruitment/function and gastric mucosal responses to aspirin.


Assuntos
Araquidonato 5-Lipoxigenase/fisiologia , Aspirina/toxicidade , Inibidores de Ciclo-Oxigenase/toxicidade , Mucosa Gástrica/efeitos dos fármacos , Isoenzimas/fisiologia , Lipoxinas , Prostaglandina-Endoperóxido Sintases/fisiologia , Acetatos/toxicidade , Acetilação , Adaptação Fisiológica , Animais , Araquidonato 5-Lipoxigenase/química , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Mucosa Gástrica/enzimologia , Mucosa Gástrica/patologia , Mucosa Gástrica/fisiologia , Ácidos Hidroxieicosatetraenoicos/biossíntese , Isoenzimas/química , Leucotrieno B4/biossíntese , Inibidores de Lipoxigenase/toxicidade , Modelos Biológicos , Prostaglandina-Endoperóxido Sintases/química , Pirróis/toxicidade , Ratos
5.
J Nephrol ; 16(2): 210-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12768067

RESUMO

BACKGROUND: In a number of experimental models of nephropathy, heparin is renoprotective because it inhibits mesangial matrix synthesis and cell proliferation; in most of these models, glomerular macrophage infiltration has a pathogenic role. We investigated the hypothesis that heparin might also be renoprotective by modulating the macrophages in various ways in the chronic puromycin glomerulosclerosis model. METHODS: We studied the effect of a 3 month course, two different dosages of a non-anticoagulant heparin by immunohistochemical evaluation of the number of macrophages (ED-1 positive cells) in glomeruli, as well as the expression and deposition of TGF-beta and latent TGF-beta binding protein in foam cells and mesangial matrix. RESULTS: The renoprotective effect of heparin in this model was confirmed by our observation of lower urine protein and albumin excretion, and a reduced glomerular sclerosis score. These effects were associated with the prevention of macrophage glomerular infiltration, and the inhibition of the TGF-beta axes in foam cells as shown by the reduction in cytoplasm immunostaining for TGF-beta and LTBP-1; heparin also reduced peri-macrophagic collagen IV deposition. CONCLUSIONS: The inhibition of TGF-beta in macrophages seems to be part of heparin general activities. The inhibitory effect of heparin on macrophage infiltration and TGF-beta synthesis in this renal disease model supports the notion that heparin and derived molecules constitute potentially useful therapeutic agents in nephropathies.


Assuntos
Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Glomerulosclerose Segmentar e Focal/patologia , Heparina/farmacologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/efeitos dos fármacos , Análise de Variância , Animais , Biópsia por Agulha , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Taxa de Filtração Glomerular , Imuno-Histoquímica , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Probabilidade , Puromicina , Ratos , Ratos Wistar , Sensibilidade e Especificidade
6.
J Antibiot (Tokyo) ; 67(9): 667-70, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25095806

RESUMO

Rifaximin is a non-systemic oral antibiotic derived from rifampin and characterized by a broad spectrum of antibacterial activity against Gram-positive and -negative, aerobic and anaerobic bacteria. Rifaximin was first approved in Italy in 1987 and afterwards in many other worldwide countries for the treatment of several gastrointestinal diseases. This review updates the pharmacology and pharmacodynamics of rifaximin highlighting the different actions, beyond its antibacterial activity, such as alteration of virulence, prevention of gut mucosal adherence and bacterial translocation. Moreover, rifaximin exerts some anti-inflammatory effects with only a minimal effect on the overall composition of the gut microbiota. All these properties make rifaximin a good candidate to treat various gastrointestinal diseases.


Assuntos
Antibacterianos/farmacologia , Infecções Bacterianas/tratamento farmacológico , Rifamicinas/farmacologia , Animais , Bactérias/efeitos dos fármacos , Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Fármacos Gastrointestinais/farmacologia , Gastroenteropatias/tratamento farmacológico , Trato Gastrointestinal/microbiologia , Humanos , Rifaximina
7.
Eur J Pharmacol ; 668(1-2): 317-24, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21806984

RESUMO

A dysregulated interaction between intestinal epithelial cells (IEC) and components of innate immunity is a hallmark of inflammatory bowel diseases. Rifaximin is a poorly absorbed oral antimicrobial agent increasingly used in the treatment of inflammatory bowel diseases that has been demonstrated to act as a gut-specific ligand for the human nuclear receptor pregnane-X receptor (PXR). In the present study we investigated, whether activation of PXR in IEC by rifaximin, emanates counter-regulatory signals and modulates the expression of cytokines or chemokines mechanistically involved in dysregulated intestinal immune homeostasis documented in inflammatory bowel diseases. Our results demonstrate that primary IEC express PXR that regulate the pattern of cytokines and chemokines expressed. PXR silencing decreases TGF-ß and IP-10 while increases the expression of TNF-α, IL-8, Rantes and increase the production of PGE2. This pattern is further exacerbated by treating anti-PXR siRNA cells with bacterial endotoxin (LPS). Exposure to rifaximin caused a robust attenuation of generation of inflammatory mediators caused by LPS and increased the generation of TGF-ß. PXR silencing completely abrogated these anti-inflammatory effects of rifaximin. By Western blot analysis we found that rifaximin abrogates the binding of NF-κB caused by LPS. Finally, exposure of human colon biopsies from inflammatory bowel diseases patients to rifaximin reduced mRNA levels of IL-8, Rantes, MIP-3α and TNFα induced by LPS. Collectively, these data establish that rifaximin exerts counter-regulatory activities at the interface between enteric bacteria and intestinal epithelial cells. The ability of rifaximin to activate PXR contributes to the maintenance of the intestinal immune homeostasis.


Assuntos
Imunidade Inata/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , NF-kappa B/antagonistas & inibidores , Receptores de Esteroides/metabolismo , Rifamicinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Quimiocinas/biossíntese , Colo/efeitos dos fármacos , Colo/imunologia , Colo/metabolismo , Colo/patologia , DNA/metabolismo , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/imunologia , Inativação Gênica , Humanos , Imunidade Inata/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Receptor de Pregnano X , Receptores de Esteroides/deficiência , Receptores de Esteroides/genética , Rifaximina , Transdução de Sinais/genética , Transdução de Sinais/imunologia
8.
Biochem Pharmacol ; 80(11): 1700-7, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20816942

RESUMO

The pregnane-X-receptor (PXR) is master gene overseeing detoxification of wide number of xenobiotics and is critical for maintenance of intestinal integrity. The intestinal expression of genes involved in cellular detoxification is down-regulated in patients with inflammatory bowel diseases (IBD). Rifaximin is a non-absorbable antibiotic endowed with a PXR agonistic activity. In the present study we have investigated whether rifaximin activates PXR in primary human colon epithelial cells and human colon biopsies and assessed whether this antibiotic antagonizes the effect of tumor necrosis factor (TNF)-α on expression of PXR and PXR-related genes. Present results demonstrate that primary colon epithelial cells express PXR and that their exposure to rifaximin induces the expression of genes involved in cellular detoxification. Exposure to TNFα reduces the expression of PXR mRNA as well as expression of its target genes. This inhibitory effect was prevented by that co-treatment with rifaximin. Knocking down the expression of PXR in colon epithelial cells by an anti-PXR siRNA, abrogated the counter-regulatory effects exerted by rifaximin on cell exposed to TNFα. Finally, ex vivo exposure of colon biopsies obtained from ulcerative colitis patients to rifaximin increased the expression of genes involved in xenobiotics metabolism. In aggregate, these data illustrate that rifaximin increases the expression of PXR and PXR-regulated genes involved in the metabolism and excretion of xenobiotics and antagonizes the effects of TNFα in intestinal epithelial cells and colon biopsies. These non-antibiotic effects of rifaximin could contribute to the maintenance of the intestinal barrier integrity against xenobiotics and products generated by luminal bacteria.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Receptores de Esteroides/fisiologia , Rifamicinas/farmacologia , Transdução de Sinais/fisiologia , Adulto , Anti-Inflamatórios não Esteroides/farmacocinética , Linhagem Celular , Colo/citologia , Colo/efeitos dos fármacos , Colo/metabolismo , Feminino , Células Hep G2 , Humanos , Inativação Metabólica/fisiologia , Mucosa Intestinal/citologia , Masculino , Receptor de Pregnano X , Rifamicinas/farmacocinética , Rifaximina , Transdução de Sinais/efeitos dos fármacos
9.
Digestion ; 66(4): 246-56, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12592101

RESUMO

BACKGROUND: A modification of the intestinal flora and an increased bacterial translocation is a common finding in patients with inflammatory bowel disease as well as in animal model of colitis. Rifaximin, a non-absorbable derivative of rifamycin, is an effective antibiotic that acts by inhibiting bacterial ribonucleic acid synthesis. AIMS: In the present study, we investigated the effect of the administration of rifaximin (10, 30 and 50 mg/kg/day) or prednisolone (10 mg/kg/day) in 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. METHODS: Colitis was induced in mice by intrarectal administration of TNBS (1.5 mg/mouse in 50% ethanol) and disease severity assessed clinically and by histologic scoring of colon damage, determination of interleukin (IL)-2, IL-12, interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha (protein and mRNA and myeloperoxidase (MPO) activity in the colon. Cytokines production by the lamina propria mononuclear cells (LPMC) and luminal bacteria were also measured. RESULTS: Rifaximin administration (30 or 50 mg/kg/day) increased survival rates of colitic mice and reduced colitis severity as demonstrated by improvement of wasting syndrome, histologic scores, decrease in colon IL-2, IL-12, IFN-gamma and TNF-alpha (protein and mRNA) levels, and diminished colon MPO activity. Rifaximin administration caused a significant reduction of colon bacterial translocation towards mesenteric lymph nodes. LPMC obtained from rifaximin-treated mice released significantly lower amount of IFN-gamma in response to ex vivo stimulation with agonistic anti-CD3 and anti-CD28 antibodies. Rifaximin (50 mg/kg/day) significantly accelerates recovery in mice with established colitis. CONCLUSIONS: Luminal bacterial microflora plays a role in the pathogenesis of TNBS-induced colitis in mice. Rifaximin administration reduces the development of colitis and accelerates healing of established disease by preventing bacterial translocation.


Assuntos
Antibacterianos/uso terapêutico , Translocação Bacteriana/efeitos dos fármacos , Colite/prevenção & controle , Mucosa Intestinal/imunologia , Leucócitos Mononucleares/imunologia , Rifamicinas/uso terapêutico , Doença Aguda , Animais , Anti-Inflamatórios/uso terapêutico , Colite/imunologia , Colite/microbiologia , Colite/patologia , Colo/imunologia , Colo/microbiologia , Colo/patologia , Citocinas/biossíntese , Relação Dose-Resposta a Droga , Interferon gama/biossíntese , Mucosa Intestinal/patologia , Linfonodos/microbiologia , Mesentério , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Peroxidase/metabolismo , Prednisolona/uso terapêutico , Rifaximina , Ácido Trinitrobenzenossulfônico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA